Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Proc Natl Acad Sci U S A ; 116(9): 3909-3918, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30733294

ABSTRACT

Metabolic plasticity enables cancer cells to switch their metabolism phenotypes between glycolysis and oxidative phosphorylation (OXPHOS) during tumorigenesis and metastasis. However, it is still largely unknown how cancer cells orchestrate gene regulation to balance their glycolysis and OXPHOS activities. Previously, by modeling the gene regulation of cancer metabolism we have reported that cancer cells can acquire a stable hybrid metabolic state in which both glycolysis and OXPHOS can be used. Here, to comprehensively characterize cancer metabolic activity, we establish a theoretical framework by coupling gene regulation with metabolic pathways. Our modeling results demonstrate a direct association between the activities of AMPK and HIF-1, master regulators of OXPHOS and glycolysis, respectively, with the activities of three major metabolic pathways: glucose oxidation, glycolysis, and fatty acid oxidation. Our model further characterizes the hybrid metabolic state and a metabolically inactive state where cells have low activity of both glycolysis and OXPHOS. We verify the model prediction using metabolomics and transcriptomics data from paired tumor and adjacent benign tissue samples from a cohort of breast cancer patients and RNA-sequencing data from The Cancer Genome Atlas. We further validate the model prediction by in vitro studies of aggressive triple-negative breast cancer (TNBC) cells. The experimental results confirm that TNBC cells can maintain a hybrid metabolic phenotype and targeting both glycolysis and OXPHOS is necessary to eliminate their metabolic plasticity. In summary, our work serves as a platform to symmetrically study how tuning gene activity modulates metabolic pathway activity, and vice versa.


Subject(s)
AMP-Activated Protein Kinases/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Metabolic Networks and Pathways/genetics , Triple Negative Breast Neoplasms/genetics , AMP-Activated Protein Kinases/metabolism , Cell Line, Tumor , Fatty Acids/metabolism , Female , Glucose/metabolism , Glycolysis/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mitochondria/metabolism , Models, Theoretical , Oxidative Phosphorylation , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
2.
Br J Cancer ; 124(12): 1902-1911, 2021 06.
Article in English | MEDLINE | ID: mdl-33859341

ABSTRACT

Cancer cells have the plasticity to adjust their metabolic phenotypes for survival and metastasis. A developmental programme known as epithelial-to-mesenchymal transition (EMT) plays a critical role during metastasis, promoting the loss of polarity and cell-cell adhesion and the acquisition of motile, stem-cell characteristics. Cells undergoing EMT or the reverse mesenchymal-to-epithelial transition (MET) are often associated with metabolic changes, as the change in phenotype often correlates with a different balance of proliferation versus energy-intensive migration. Extensive crosstalk occurs between metabolism and EMT, but how this crosstalk leads to coordinated physiological changes is still uncertain. The elusive connection between metabolism and EMT compromises the efficacy of metabolic therapies targeting metastasis. In this review, we aim to clarify the causation between metabolism and EMT on the basis of experimental studies, and propose integrated theoretical-experimental efforts to better understand the coupled decision-making of metabolism and EMT.


Subject(s)
Energy Metabolism/physiology , Epithelial-Mesenchymal Transition/physiology , Neoplasms/pathology , Animals , Cell Differentiation , Epithelial-Mesenchymal Transition/genetics , Humans , Neoplasm Metastasis , Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/physiology
3.
Asian-Australas J Anim Sci ; 31(3): 457-466, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29268577

ABSTRACT

OBJECTIVE: The present study aimed to assess the nitrogen (N) use efficiency of acidified pig slurry for regrowth yield and its environmental impacts on perennial ryegrass swards. METHODS: The pH of digested pig slurry was adjusted to 5.0 or 7.0 by the addition of sulfuric acid and untreated as a control. The pig slurry urea of each treatment was labeled with 15N urea and applied at a rate of 200 kg N/ha immediately after cutting. Soil and herbage samples were collected at 7, 14, and 56 d of regrowth. The flux of pig slurry-N to regrowth yield and soil N mineralization were analyzed, and N losses via NH3, N2O emission and NO3- leaching were also estimated. RESULTS: The pH level of the applied slurry did not have a significant effect on herbage yield or N content of herbage at the end of regrowth, whereas the amount of N derived from pig slurry urea (NdfSU) was higher in both herbage and soils in pH-controlled plots. The NH4+-N content and the amount of N derived from slurry urea into soil NH4+ fraction (NdfSU-NH4+) was significantly higher in in the pH 5 plot, whereas NO3- and NdfSU-NO3- were lower than in control plots over the entire regrowth period. Nitrification of NH4+-N was delayed in soil amended with acidified slurry. Compared to non-pH-controlled pig slurry (i.e. control plots), application of acidified slurry reduced NH3 emissions by 78.1%, N2O emissions by 78.9% and NO3- leaching by 17.81% over the course of the experiment. CONCLUSION: Our results suggest that pig slurry acidification may represent an effective means of minimizing hazardous environmental impacts without depressing regrowth yield.

4.
Hepatology ; 63(3): 864-79, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26599259

ABSTRACT

UNLABELLED: The death rate from hepatocellular carcinoma (HCC) is increasing, and liver cancer is the second leading cause of cancer-related mortality worldwide. Most patients with HCC have underlying liver cirrhosis and compromised liver function, limiting treatment options. Cirrhosis is associated with cell dedifferentiation and expansion of hepatocholangiolar progenitor cells. We identified a microRNA signature associated with HCC and hepatocytic differentiation of progenitor cells. We further identified miR-148a as an inducer of hepatocytic differentiation that is down-regulated in HCC. MiR-148a-mimetic treatment in vivo suppressed tumor growth, reduced tumor malignancy and liver fibrosis, and prevented tumor development. These effects were associated with an increased differentiated phenotype and mediated by IκB kinase alpha/NUMB/NOTCH signaling. CONCLUSION: miR-148a is an inhibitor of the IκB kinase alpha/NUMB/NOTCH pathway and an inducer of hepatocytic differentiation that when deregulated promotes HCC initiation and progression. Differentiation-targeted therapy may be a promising strategy to treat and prevent HCC.


Subject(s)
Carcinoma, Hepatocellular/therapy , Cell Differentiation , Liver Neoplasms/therapy , MicroRNAs/metabolism , Animals , Benzazepines , Biomarkers/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Fibrosis , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , I-kappa B Kinase/metabolism , Liver/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Receptors, Notch/antagonists & inhibitors , Receptors, Notch/metabolism
5.
Stem Cells ; 34(5): 1284-96, 2016 05.
Article in English | MEDLINE | ID: mdl-26731713

ABSTRACT

MicroRNA expression profiling in human liver progenitor cells following hepatocytic differentiation identified miR-122 and miR-194 as the microRNAs most strongly upregulated during hepatocytic differentiation of progenitor cells. MiR-194 was also highly upregulated following hepatocytic differentiation of human embryonic stem cells (hESCs). Overexpression of miR-194 in progenitor cells accelerated their differentiation into hepatocytes, as measured by morphological features such as canaliculi and expression of hepatocytic markers. Overexpression of miR-194 in hESCs induced their spontaneous differentiation, a phenotype accompanied with accelerated loss of the pluripotent factors OCT4 and NANOG and decrease in mesoderm marker HAND1 expression. We then identified YAP1 as a direct target of miR-194. Inhibition of YAP1 strongly induced hepatocytic differentiation of progenitor cells and YAP1 overexpression reversed the miR-194-induced hepatocytic differentiation of progenitor cells. In conclusion, we identified miR-194 as a potent inducer of hepatocytic differentiation of progenitor cells and further identified YAP1 as a mediator of miR-194's effects on hepatocytic differentiation and liver progenitor cell fate. Stem Cells 2016;34:1284-1296.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Differentiation/genetics , Hepatocytes/cytology , Hepatocytes/metabolism , MicroRNAs/metabolism , Phosphoproteins/metabolism , Base Sequence , Cell Line , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Humans , Liver/cytology , MicroRNAs/genetics , Transcription Factors , Up-Regulation/genetics , YAP-Signaling Proteins
6.
J Hepatol ; 63(2): 408-19, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25817558

ABSTRACT

BACKGROUND & AIMS: Most common reason behind changes in histone deacetylase (HDAC) function is its overexpression in cancer. However, among HDACs in liver cancer, HDAC6 is uniquely endowed with a tumor suppressor, but the mechanism underlying HDAC6 inactivation has yet to be uncovered. METHODS: Microarray profiling and target prediction programs were used to identify miRNAs targeting HDAC6. A series of inhibitors, activators and siRNAs was introduced to validate regulatory mechanisms for microRNA-221-3p (miR-221) governing HDAC6 in hepatocarcinogenesis. RESULTS: Comprehensive miRNA profiling analysis identified seven putative endogenous miRNAs that are significantly upregulated in hepatocellular carcinoma (HCC). While miR-221 was identified as a suppressor of HDAC6 by ectopic expression of miRNA mimics in Dicer knockdown cells, targeted-disruption of miR-221 repressed cancer cell growth through derepressing HDAC6 expression. Suppression of HDAC6 via miR-221 was induced by JNK/c-Jun signaling in liver cancer cells but not in normal hepatic cells. Additionally, cytokine-induced NF-κBp65 independently regulated miR-221, thereby suppressing HDAC6 expression in HCC cells. HCC tissues derived from chemical-induced rat and H-ras12V transgenic mice liver cancer models validated that JNK/c-Jun activation and NF-κBp65 nuclear translocation are essential for the transcription of miR-221 leading to repression of HDAC6 in HCC. CONCLUSIONS: Our findings suggest that the functional loss or suppression of the tumor suppressor HDAC6 is caused by induction of miR-221 through coordinated JNK/c-Jun- and NF-κB-signaling pathways during liver tumorigenesis, providing a novel target for the molecular treatment of liver malignancies.


Subject(s)
Gene Expression Regulation, Neoplastic , Histone Deacetylases/genetics , Liver Neoplasms, Experimental/genetics , MicroRNAs/genetics , RNA, Neoplasm/genetics , Animals , Disease Progression , Histone Deacetylase 6 , Histone Deacetylases/biosynthesis , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , MicroRNAs/biosynthesis , Polymerase Chain Reaction , Rats
7.
Hepatology ; 57(3): 1055-67, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23079745

ABSTRACT

UNLABELLED: Sirtuins are nicotinamide adenine dinucleotide oxidized form (NAD(+) )-dependent deacetylases and function in cellular metabolism, stress resistance, and aging. For sirtuin7 (SIRT7), a role in ribosomal gene transcription is proposed, but its function in cancer has been unclear. In this study we show that SIRT7 expression was up-regulated in a large cohort of human hepatocellular carcinoma (HCC) patients. SIRT7 knockdown influenced the cell cycle and caused a significant increase of liver cancer cells to remain in the G1 /S phase and to suppress growth. This treatment restored p21(WAF1/Cip1) , induced Beclin-1, and repressed cyclin D1. In addition, sustained suppression of SIRT7 reduced the in vivo tumor growth rate in a mouse xenograft model. To explore mechanisms in SIRT7 regulation, microRNA (miRNA) profiling was carried out. This identified five significantly down-regulated miRNAs in HCC. Bioinformatics analysis of target sites and ectopic expression in HCC cells showed that miR-125a-5p and miR-125b suppressed SIRT7 and cyclin D1 expression and induced p21(WAF1/Cip1) -dependent G1 cell cycle arrest. Furthermore, treatment of HCC cells with 5-aza-2'-deoxycytidine or ectopic expression of wildtype but not mutated p53 restored miR-125a-5p and miR-125b expression and inhibited tumor cell growth, suggesting their regulation by promoter methylation and p53 activity. To show the clinical significance of these findings, mutations in the DNA binding domain of p53 and promoter methylation of miR-125b were investigated. Four out of nine patients with induced SIRT7 carried mutations in the p53 gene and one patient showed hypermethylation of the miR-125b promoter region. CONCLUSION: Our findings suggest the oncogenic potential of SIRT7 in hepatocarcinogenesis. A regulatory loop is proposed whereby SIRT7 inhibits transcriptional activation of p21(WAF1/Cip1) by way of repression of miR-125a-5p and miR-125b. This makes SIRT7 a promising target in cancer therapy. (HEPATOLOGY 2013).


Subject(s)
Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Neoplastic/genetics , Liver Neoplasms/genetics , MicroRNAs/genetics , Sirtuins/genetics , Animals , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Gene Knockdown Techniques , Genetic Therapy/methods , Genomics , Humans , Liver Neoplasms/metabolism , Mice , Mice, Nude , RNA, Messenger/metabolism , Sirtuins/metabolism , Xenograft Model Antitumor Assays
8.
Cancer Lett ; 587: 216724, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38373689

ABSTRACT

CD24 is a well-characterized breast cancer (BC) stem cell (BCSC) marker. Primary breast tumor cells having CD24-negativity together with CD44-positivity is known to maintain high metastatic potential. However, the functional role of CD24 gene in triple-negative BC (TNBC), an aggressive subtype of BC, is not well understood. While the significance of CD24 in regulating immune pathways is well recognized in previous studies, the significance of CD24 low expression in onco-signaling and metabolic rewiring is largely unknown. Using CD24 knock-down and over-expression TNBC models, our in vitro and in vivo analysis suggest that CD24 is a tumor suppressor in metastatic TNBC. Comprehensive in silico gene expression analysis of breast tumors followed by lipidomic and metabolomic analyses of CD24-modulated cells revealed that CD24 negativity induces mitochondrial oxidative phosphorylation and reprograms TNBC metabolism toward the fatty acid beta-oxidation (FAO) pathway. CD24 silencing activates PPARα-mediated regulation of FAO in TNBC cells. Further analysis using reverse-phase protein array and its validation using CD24-modulated TNBC cells and xenograft models nominated CD24-NF-κB-CPT1A signaling pathway as the central regulatory mechanism of CD24-mediated FAO activity. Overall, our study proposes a novel role of CD24 in metabolic reprogramming that can open new avenues for the treatment strategies for patients with metastatic TNBC.


Subject(s)
NF-kappa B , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/pathology , PPAR alpha/genetics , Cell Line, Tumor , Fatty Acids/metabolism , CD24 Antigen/genetics , CD24 Antigen/metabolism
9.
Hepatology ; 56(2): 644-57, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22392728

ABSTRACT

UNLABELLED: Ubiquitin-binding histone deacetylase 6 (HDAC6) is uniquely endowed with tubulin deacetylase activity and plays an important role in the clearance of misfolded protein by autophagy. In cancer, HDAC6 has become a target for drug development due to its major contribution to oncogenic cell transformation. In the present study we show that HDAC6 expression was down-regulated in a large cohort of human hepatocellular carcinoma (HCC) patients, and that low expression of HDAC6 was significantly associated with poor prognosis of HCC patients in 5-year overall, disease-free, and recurrence-free survival. Notably, we observed that ectopic overexpression of HDAC6 suppressed tumor cell growth and proliferation in various liver cancer cells, and elicited increased LC3B-II conversion and autophagic vacuole formation without causing apoptotic cell death or cell cycle inhibition. In addition, the sustained overexpression of HDAC6 reduced the in vivo tumor growth rate in a mouse xenograft model. It was also found that HDAC6 mediated autophagic cell death by way of Beclin 1 and activation of the LC3-II pathway in liver cancer cells, and that HDAC6 overexpression activated c-Jun NH2-terminal kinase (JNK) and increased the phosphorylation of c-Jun. In contrast, the induction of Beclin 1 expression was blocked by SP600125 (a specific inhibitor of JNK) or by small interfering RNA directed against HDAC6. CONCLUSION: Our findings suggest that loss of HDAC6 expression in human HCCs and tumor suppression by HDAC6 occur by way of activation of caspase-independent autophagic cell death through the JNK/Beclin 1 pathway in liver cancer and, thus, that a novel tumor suppressor function mechanism involving HDAC6 may be amenable to nonepigenetic regulation.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Autophagy/physiology , Carcinoma, Hepatocellular , Histone Deacetylases/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Liver Neoplasms , Membrane Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Beclin-1 , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Gene Expression Regulation, Neoplastic/physiology , Genes, Tumor Suppressor/physiology , Hep G2 Cells , Histone Deacetylase 6 , Histone Deacetylases/genetics , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Membrane Proteins/genetics , Mice , Mice, Nude , Neoplasm Transplantation , Prognosis , RNA, Small Interfering/genetics , Transplantation, Heterologous
10.
J Cell Biochem ; 113(6): 2167-77, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22492270

ABSTRACT

Histone deacetylase 2 (HDAC2) is crucial for embryonic development, affects cytokine signaling relevant for immune responses, and is often significantly overexpressed in solid tumors, but little is known of its role in human lung cancer. In this study, we demonstrated the aberrant expression of HDAC2 in lung cancer tissues and investigated oncogenic properties of HDAC2 in human lung cancer cell lines. HDAC2 inactivation resulted in regression of tumor cell growth and activation of cellular apoptosis via p53 and Bax activation and Bcl2 suppression. In cell cycle regulation, HDAC2 inactivation caused induction of p21WAF1/CIP1 expression, and simultaneously suppressed the expressions of cyclin E2, cyclin D1, and CDK2, respectively. Consequently, this led to the hypophosphorylation of pRb protein in G1/S transition and thereby inactivated E2F/DP1 target gene transcriptions of A549 cells. In addition, we demonstrated that HDAC2 directly regulated p21WAF1/CIP1 expression in a p53-independent manner. However, HDAC1 was not related to p21WAF1/CIP1 expression and tumorigenesis of lung cancer. Lastly, we observed that sustained-suppression of HDAC2 in A549 lung cancer cells attenuated in vitro tumorigenic properties and in vivo tumor growth of the mouse xenograft model. Taken together, we suggest that the aberrant regulation of HDAC2 and its epigenetic regulation of gene transcription in apoptosis and cell cycle components play an important role in the development of lung cancer.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Cell Cycle Proteins/metabolism , Histone Deacetylase 2/metabolism , Lung Neoplasms/metabolism , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cyclin D1/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclins/metabolism , E2F Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , Mice , Mice, Nude , Phosphorylation , RNA Interference , RNA, Small Interfering , Retinoblastoma Protein/metabolism , Transcription, Genetic , Transplantation, Heterologous , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism , bcl-Associated Death Protein/metabolism
11.
Environ Sci Technol ; 46(23): 12882-9, 2012 Dec 04.
Article in English | MEDLINE | ID: mdl-23153324

ABSTRACT

Persistent organic pollutants (POPs) are degradation-resistant anthropogenic chemicals that accumulate in the food chain and in adipose tissue, and are among the most hazardous compounds ever synthesized. However, their toxic mechanisms are still undefined. To investigate whether characteristic molecular signatures can discriminate individual POP and provide prediction markers for the early detection of POPs exposure in an animal model, we performed transcriptomic analysis of rat liver tissues after exposure to POPs. The six different POPs (toxaphene, hexachlorobenzene, chlordane, mirex, dieldrin, and heptachlor) were administered to 11-week-old male Sprague-Dawley rats, and after 48 h of exposure, RNAs were extracted from liver tissues and subjected to rat whole genome expression microarrays. Early during exposure, conventional toxicological analysis including changes in the body and organ weight, histopathological examination, and blood biochemical analysis did not reflect any toxicant stresses. However, unsupervised gene expression analysis of rat liver tissues revealed in a characteristic molecular signature for each toxicant, and supervised analysis identified 2708 outlier genes that discerned the POPs exposure group from the vehicle-treated control. Combination analysis of two different multiclassifications suggested 384 genes as early detection markers for predicting each POP exposure with 100% accuracy. The data from large-scale gene expression analysis of a different POP exposure in rat model suggest that characteristic expression profiles exist in liver hepatic cells and multiclassification of POP-specific molecular signatures can discriminate each toxicant at an early exposure time. The use of these molecular markers may be more widely implemented in combination with more traditional techniques for assessment and prediction of toxicity exposure to POPs from an environmental aspect.


Subject(s)
Chemical and Drug Induced Liver Injury/diagnosis , Environmental Pollutants/toxicity , Liver/drug effects , Pesticides/toxicity , Transcriptome/drug effects , Animals , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/pathology , Dieldrin/toxicity , Fungicides, Industrial/toxicity , Heptachlor/toxicity , Hexachlorobenzene/toxicity , Insecticides/toxicity , Liver/metabolism , Liver/pathology , Male , Mirex/toxicity , Rats , Rats, Sprague-Dawley , Toxaphene/toxicity , Toxicity Tests/methods
12.
Commun Biol ; 5(1): 493, 2022 05 24.
Article in English | MEDLINE | ID: mdl-35610507

ABSTRACT

The major limitations of DNA-targeting chemotherapy drugs include life-threatening toxicity, acquired resistance and occurrence of secondary cancers. Here, we report a small molecule, Carbazole Blue (CB), that binds to DNA and inhibits cancer growth and metastasis by targeting DNA-related processes that tumor cells use but not the normal cells. We show that CB inhibits the expression of pro-tumorigenic genes that promote unchecked replication and aberrant DNA repair that cancer cells get addicted to survive. In contrast to chemotherapy drugs, systemic delivery of CB suppressed breast cancer growth and metastasis with no toxicity in pre-clinical mouse models. Using PDX and ex vivo explants from estrogen receptor (ER) positive, ER mutant and TNBC patients, we further demonstrated that CB effectively blocks therapy-sensitive and therapy-resistant breast cancer growth without affecting normal breast tissue. Our data provide a strong rationale to develop CB as a viable therapeutic for treating breast cancers.


Subject(s)
Breast Neoplasms , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , DNA , DNA Repair , Female , Humans , Mice , Receptors, Estrogen/metabolism
13.
Toxicol Appl Pharmacol ; 250(2): 162-9, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-20955722

ABSTRACT

In a previous report we demonstrated that the transcriptomic response of liver tissue was specific to toxicants, and a characteristic molecular signature could be used as an early prognostic biomarker in rats. It is necessary to determine the transcriptomic response to toxicants in peripheral blood for application to the human system. Volatile organic compounds (VOCs) comprise a major group of pollutants which significantly affect the chemistry of the atmosphere and human health. In this study we identified and validated the specific molecular signatures of toxicants in rat whole blood as early predictors of environmental toxicants. VOCs (dichloromethane, ethylbenzene, and trichloroethylene) were administered to 11-week-old SD male rats after 48h of exposure, peripheral whole blood was subjected to expression profiling analysis. Unsupervised gene expression analysis resulted in a characteristic molecular signature for each toxicant, and supervised analysis identified 1,217 outlier genes as distinct molecular signatures discerning VOC exposure from healthy controls. Further analysis of multi-classification suggested 337 genes as early detective molecular markers for three VOCs with 100% accuracy. A large-scale gene expression analysis of a different VOC exposure animal model suggested that characteristic expression profiles exist in blood cells and multi-classification of this VOC-specific molecular signature can discriminate each toxicant at an early exposure time. This blood expression signature can thus be used as discernable surrogate marker for detection of biological responses to VOC exposure in an environment.


Subject(s)
Benzene Derivatives/toxicity , Gene Expression Regulation/drug effects , Methylene Chloride/toxicity , Trichloroethylene/toxicity , Animals , Biomarkers/metabolism , Environmental Exposure/adverse effects , Gene Expression Profiling , Male , Rats , Rats, Sprague-Dawley , Time Factors
14.
Environ Sci Technol ; 45(1): 300-6, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21133357

ABSTRACT

Whole blood is one of the most easily accessible biofluids, and circulating leukocytes would include informative transcripts as a first line of immune defense for many disease processes. To demonstrate that transcriptomic responses of circulating blood cells reflect the exposure to environmental toxicants and the characteristic molecular signatures can discriminate and predict the type of toxicant at an early exposure time, we identified and validated characteristic gene expression profiles of rat whole blood after exposure to polycyclic aromatic hydrocarbons (PAHs). At an early exposure time point, conventional toxicological analysis including changes in the body and organ weight, histopathological examination, and blood biochemical analysis did not reflect any toxicant stresses. However, unsupervised gene expression analysis of blood cells resulted in a characteristic molecular signature for each toxicant. Further analysis of multiclassification suggested 220 genes as early detective and surrogate markers for predicting each PAH with 100% accuracy. These findings suggest that the blood expression signature could be used as a predictable and discernible surrogate marker for detection and prediction of PAHs, and the use of these molecular markers may be more widely implemented in combination with more traditional techniques for assessment and prediction of toxicity exposure to PAHs from an environmental aspect.


Subject(s)
Environmental Exposure/analysis , Environmental Pollutants/blood , Polycyclic Aromatic Hydrocarbons/blood , Animals , Benz(a)Anthracenes/blood , Benz(a)Anthracenes/toxicity , Benzo(a)pyrene/metabolism , Benzo(a)pyrene/toxicity , Biomarkers/blood , Environmental Exposure/statistics & numerical data , Environmental Pollutants/toxicity , Gene Expression/drug effects , Gene Expression Profiling , Male , Naphthalenes/blood , Naphthalenes/toxicity , Phenanthrenes/blood , Phenanthrenes/toxicity , Polycyclic Aromatic Hydrocarbons/toxicity , Rats , Rats, Sprague-Dawley
15.
J Cell Biochem ; 110(3): 687-96, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20512928

ABSTRACT

The Wnt/beta-catenin signaling pathway regulates various aspects of development and plays important role in human carcinogenesis. Nemo-like kinase (NLK), which is mediator of Wnt/beta-catenin signaling pathway, phosphorylates T-cell factor/lymphoid enhancer factor (TCF/LEF) factor and inhibits interaction of beta-catenin/TCF complex. Although, NLK is known to be a tumor suppressor in Wnt/beta-catenin signaling pathway of colon cancer, the other events occurring downstream of NLK pathways in other types of cancer remain unclear. In the present study, we identified that expression of NLK was significantly up-regulated in the HCCs compared to corresponding normal tissues in five selected tissue samples. Immunohistochemical analysis showed significant over-expression of NLK in the HCCs. Targeted-disruption of NLK suppressed cell growth and arrested cell cycle transition. Suppression of NLK elicited anti-mitogenic properties of the Hep3B cells by simultaneous inhibition of cyclinD1 and CDK2. The results of this study suggest that NLK is aberrantly regulated in HCC, which might contribute to the mitogenic potential of tumor cells during the initiation and progression of hepatocellular carcinoma; this process appears to involve the induction of CDK2 and cyclin D1 and might provide a novel target for therapeutic intervention in patients with liver cancer.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cyclin D1/metabolism , Cyclin-Dependent Kinase 2/metabolism , Liver Neoplasms/metabolism , Transcription Factor TFIIIA/metabolism , Apoptosis/physiology , Blotting, Western , Carcinoma, Hepatocellular/genetics , Cell Cycle/physiology , Cell Cycle Proteins , Cell Proliferation , Cyclin D1/genetics , Cyclin-Dependent Kinase 2/genetics , Gene Expression , Gene Expression Regulation , Gene Silencing , Humans , Immunohistochemistry , Liver Neoplasms/genetics , Membrane Transport Proteins , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Transcription Factor TFIIIA/genetics
16.
Toxicol Appl Pharmacol ; 237(1): 91-101, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19285098

ABSTRACT

The amphetamine derivative (+/-)-3,4-methylenedioxymethamphetamine (MDMA or ecstasy) is a synthetic amphetamine analogue used recreationally to obtain an enhanced affiliative emotional response. MDMA is a potent monoaminergic neurotoxin with the potential to damage brain serotonin and/or dopamine neurons. As the majority of MDMA users are young adults, the risk that users may expose the fetus to MDMA is a concern. However, the majority of studies on MDMA have investigated the effects on adult animals. Here, we investigated whether long-term exposure to MDMA, especially in adolescence, could induce comprehensive transcriptional changes in mouse brain. Transcriptomic analysis of mouse brain regions demonstrated significant gene expression changes in the cerebral cortex. Supervised analysis identified 1028 genes that were chronically dysregulated by long-term exposure to MDMA in adolescent mice. Functional categories most represented by this MDMA characteristic signature are intracellular molecular signaling pathways of neurotoxicity, such as, the MAPK signaling pathway, the Wnt signaling pathway, neuroactive ligand-receptor interaction, long-term potentiation, and the long-term depression signaling pathway. Although these resultant large-scale molecular changes remain to be studied associated with functional brain damage caused by MDMA, our observations delineate the possible neurotoxic effects of MDMA on brain function, and have therapeutic implications concerning neuro-pathological conditions associated with MDMA abuse.


Subject(s)
Cerebral Cortex/metabolism , Hallucinogens/toxicity , N-Methyl-3,4-methylenedioxyamphetamine/toxicity , Nerve Tissue Proteins/metabolism , Synaptic Transmission/drug effects , Age Factors , Analysis of Variance , Animals , Cerebellum/drug effects , Cerebellum/growth & development , Cerebellum/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/growth & development , Critical Period, Psychological , Female , Gene Expression Profiling , Hippocampus/drug effects , Hippocampus/growth & development , Hippocampus/metabolism , Longitudinal Studies , Male , Mesencephalon/drug effects , Mesencephalon/growth & development , Mesencephalon/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/drug effects , Pons/drug effects , Pons/growth & development , Pons/metabolism , Signal Transduction/drug effects , Statistics, Nonparametric
17.
Oncol Rep ; 22(3): 475-80, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19639191

ABSTRACT

The transforming growth factor receptor III (TGFbetaRIII) is the most abundant and essential TGF-beta binding protein that functions as a co-receptor with other receptors in TGF-beta signaling. In earlier studies, expression of TGFbetaRIII was reported to be decreased in a variety of human cancers. Functional assessment of TGFbetaRIII was performed in many previously studied cancers but not in hepatocellular carcinoma. Therefore, in this study, we investigated the expression and genetic alterations of TGFbetaRIII in hepatocellular carcinoma (HCC) by quantitative real-time PCR (qRT-PCR) and single-strand conformation polymorphism (SSCP) analysis. The qRT-PCR showed down-regulation of TGFbetaRIII in the tumor samples. To investigate whether genetic alterations mediated decreased expression of TGFbetaRIII, we performed mutation analysis of 67 human HCC tissues by SSCP and direct sequencing. We found five previously reported and one novel single nucleotide polymorphisms in exons 2, 3, 5, 13 and 14, but no mutations were detected. These polymorphisms were not associated with amino acid changes except for a base change found in exon 2 (TCC-->TTC, S15F). The loss of heterozygosity (LOH) analysis performed on 10 tumors and corresponding normal pairs, showed a low rate of LOH (2/10). The results of this study suggest that TGFbetaRIII is transcriptionally down-regulated in hepatocellular carcinoma. In addition, genetic alterations did not appear to be associated with the reduced expression level of TGFbetaRIII. To clarify the role of TGFbetaRIII in hepatocellular tumor development and progression, functional analysis is needed in future studies.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Loss of Heterozygosity , Proteoglycans/genetics , Receptors, Transforming Growth Factor beta/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , Down-Regulation , Female , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Signal Transduction , Transforming Growth Factor beta/physiology
18.
Int J Mol Med ; 24(2): 205-26, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19578794

ABSTRACT

Histone deacetylase (HDAC) inhibitors are emerging as an exciting new class of potential anti-cancer agents for the treatment of solid and hematological malignancies. However, the best characterized HDAC function concerns the control of gene expression via the regulation of transcription activation or repression. To understand the genome-wide effects of HDAC inhibition on gene regulation, we performed serial gene expression analyses from 0 to 48 h after treating MDA-MB-435, a melanoma-derived highly metastatic tumor cell line, with Apicidin, a HDAC inhibitor. Combined-transcriptomic analysis of large-scale molecular changes induced by Apicidin resulted in the identification of 631 outlier genes that were continuously up- or down-regulated during the 48 h study period. When the 631 outlier genes were mapped to known biological processes, cell-cycle suppression emerged as the function most elicited by Apicidin. In addition comprehensive negative cell-cycle regulation by Apicidin was dissected using gene expression data and validated by Western blot analysis. We suggest the 631 outlier genes as a characteristic molecular signature for Apicidin, and propose concurrent transcriptional suppression of major components of cell-cycle regulatory circuit as potent anti-tumor mechanism of Apicidin. Genetic elements identified during this study also provide the possibility of novel therapeutic interventions in tumor metastasis.


Subject(s)
Apoptosis/drug effects , Cell Cycle/drug effects , Histone Deacetylase Inhibitors , Peptides, Cyclic/pharmacology , Apoptosis/genetics , Blotting, Western , Cell Cycle/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation/drug effects , Histone Deacetylases/metabolism , Humans , Models, Biological , Oligonucleotide Array Sequence Analysis , Smad3 Protein/genetics , Smad3 Protein/metabolism , Time Factors
19.
Cancers (Basel) ; 11(10)2019 Sep 30.
Article in English | MEDLINE | ID: mdl-31575084

ABSTRACT

Yes-associated protein (YAP)-1 is highly upregulated in pancreatic cancer and associated with tumor progression. However, little is known about the role of YAP1 and related genes in pancreatic cancer. Here, we identified target genes regulated by YAP1 and explored their role in pancreatic cancer progression and the related clinical implications. Analysis of different pancreatic cancer databases showed that Neuromedin U (NMU) expression was positively correlated with YAP1 expression in the tumor group. The Cancer Genome Atlas data indicated that high YAP1 and NMU expression levels were associated with poor mean and overall survival. YAP1 overexpression induced NMU expression and transcription and promoted cell motility in vitro and tumor metastasis in vivo via upregulation of epithelial-mesenchymal transition (EMT), whereas specific inhibition of NMU in cells stably expressing YAP1 had the opposite effect in vitro and in vivo. To define this functional association, we identified a transcriptional enhanced associate domain (TEAD) binding site in the NMU promoter and demonstrated that YAP1-TEAD binding upstream of the NMU gene regulated its transcription. These results indicate that the identified positive correlation between YAP1 and NMU is a potential novel drug target and biomarker in metastatic pancreatic cancer.

20.
Toxicology ; 249(2-3): 176-83, 2008 Jul 30.
Article in English | MEDLINE | ID: mdl-18579281

ABSTRACT

Predicting the potential human health risk posed by chemical stressors has long been a major challenge for toxicologists, and the use of microarrays to measure responses to toxicologically relevant genes, and to identify selective, sensitive biomarkers of toxicity is a major application of predictive and discovery toxicology. To investigate this possibility, we investigated whether carcinogens (at doses known to induce liver tumors in chronic exposure bioassays) deregulate characteristic sets of genes in mice. Male C3H/He mice were dosed with two hepatocarcinogens (vinyl chloride (VC, 50-25 mg/kg), aldrin (AD, 0.8-0.4 mg/kg)), or two non-hepatocarcinogens (copper sulfate (CS, 150-60 mg/kg), 2,4,5-trichlorophenoxyacetic acid (2,4,5-T, 150-60 mg/kg)). Large-scale molecular changes elicited by these four hepatotoxicants in liver tissues were analyzed using DNA microarray. Three days after administration, no significant phenotypic changes were induced by these four different hepatotoxicants in terms of histological examination or blood biochemical assay. However, unsupervised hierarchical analysis of gene expressional changes induced by hepatotoxicants resulted in two major gene subclusters on dendrogram, i.e., a carcinogen (VN, AD) and non-carcinogen group (CS, 2,4,5-T), and also revealed that distinct molecular signatures exist. These signatures were founded on well-defined functional gene categories and may differentiate genotoxic and non-genotoxic carcinogens. Furthermore, Venn diagram analysis allowed us to identify carcinogen and non-carcinogen-associated molecular signatures. Using statistical methods, we analyzed outlier genes for four different classes (genotoxic-, non-genotoxic-carcinogen, genotoxic-, non-genotoxic non-carcinogen) in terms of their potential to predict different modes-of-action. In conclusion, the identification of large-scale molecular changes in different hepatocarcinogen exposure models revealed that different types of hepatotoxicants are associated with different epigenetic changes and molecular pathways and that these large-scale characteristic molecular changes could be used as predictable toxicity markers.


Subject(s)
Chemical and Drug Induced Liver Injury/pathology , 2,4,5-Trichlorophenoxyacetic Acid/toxicity , Aldrin/toxicity , Animals , Carcinogens/toxicity , Copper Sulfate/toxicity , Data Interpretation, Statistical , Gene Expression Profiling , In Situ Hybridization , Insecticides/toxicity , Liver Neoplasms/chemically induced , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C3H , Microarray Analysis , Organ Size/drug effects , RNA/biosynthesis , RNA/genetics , Vinyl Chloride/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL